Lung dendritic cells imprint T cell lung homing and promote lung immunity through the chemokine receptor CCR4

Zamaneh Mikhak, James P Strassner, Andrew D Luster, Zamaneh Mikhak, James P Strassner, Andrew D Luster

Abstract

T cell trafficking into the lung is critical for lung immunity, but the mechanisms that mediate T cell lung homing are not well understood. Here, we show that lung dendritic cells (DCs) imprint T cell lung homing, as lung DC-activated T cells traffic more efficiently into the lung in response to inhaled antigen and at homeostasis compared with T cells activated by DCs from other tissues. Consequently, lung DC-imprinted T cells protect against influenza more effectively than do gut and skin DC-imprinted T cells. Lung DCs imprint the expression of CCR4 on T cells, and CCR4 contributes to T cell lung imprinting. Lung DC-activated, CCR4-deficient T cells fail to traffic into the lung as efficiently and to protect against influenza as effectively as lung DC-activated, CCR4-sufficient T cells. Thus, lung DCs imprint T cell lung homing and promote lung immunity in part through CCR4.

Figures

Figure 1.
Figure 1.
Lung DC–activated T cells home efficiently into the lung in response to inhaled antigen. DCs isolated from Flt3L-expanded C57BL/6 mice were used to activate Thy1.1+ OTII cells in vitro. DC-activated OTII cells were adoptively transferred into separate Thy1.2+ C57BL/6 recipient mice, followed by three inhaled OVA challenges. (a) BAL from recipients of DC-activated OTII cells were analyzed for Thy1.1+ (y axis) versus Thy1.2+ (x axis) expression in the CD4+ gate. (b) Thy1.1+ OTII cells in the BAL and lung from recipients of DC-activated T cells were enumerated 24 h after the last OVA challenge. n = 8–38 mice per group total from 2–10 independent experiments for a and b. P-values are calculated between recipients of lung DC– versus other DC-activated T cells. (c) Flow cytometry of lung DCs isolated from Flt3L-expanded mice gating on live CD11c+ cells, demonstrating the expression of CD11c (y axis) versus autofluorescence (AF) in the FITC open channel (x axis). Data are representative of three independent experiments. (d) Thy1.1+ OTII cells in the spleen and PPs from recipients of DC-activated T cells were enumerated. n = 2–3 independent experiments. (e) Lung tissue from recipients of lung DC– versus skin DC–activated OTII cells were stained with H&E and scored by histology. n = 9–10 mice per group total from three independent experiments. Bars, 150 µm. (f) DCs isolated from unexpanded C57BL/6 mice were used to activate Thy1.1+ OTII cells in vitro, which were then adoptively transferred into separate Thy1.2+ C57BL/6 recipient mice, followed by three inhaled OVA challenges. Thy1.1+ OTII cells in the BAL and lung from recipients of DC-activated OTII cells were enumerated. n = 9–23 mice per group total from three to six independent experiments. (g) DCs isolated from Flt3L-expanded C57BL/6 mice were used to activate Thy1.2+ OTI cells in vitro. DC-activated OTI cells were adoptively transferred into separate Thy1.1+ C57BL/6 recipient mice, followed by three inhaled OVA challenges. Thy1.2+ OTI cells in the BAL and lung from recipients of DC-activated OTI cells were enumerated. n = 6–15 mice per group total from two to four independent experiments. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ****, P < 0.00005. Data are presented as mean (±SEM).
Figure 2.
Figure 2.
The homing advantage of lung DC–activated T cells in response to antigen is specific to the lung. DCs isolated from Flt3L-expanded C57BL/6 mice were used to activate OTII cells in vitro. DC-activated T cells were adoptively transferred into separate recipient mice, followed by oral, i.p., or epicutaneous OVA challenges. DC-activated Thy1.1+ OTII cells were transferred into Thy1.2+ C57BL/6 recipients in a–c. DC-activated Thy1.2+ OTII cells were transferred into Thy1.1+ C57BL/6 recipients in d. (a) Thy1.1+ OTII cells in LP, PPs, lung, and spleen from recipients of lung DC– versus MLN DC–activated T cells were enumerated 24 h after one oral OVA challenge. (b) PPs from recipients of lung DC– versus MLN DC–activated T cells after one oral OVA challenge were analyzed for the expression of CD4+ (y axis) and Thy1.1+ (x axis, right) by flow cytometry. (c) Thy1.1+ OTII cells in PPs, lung, and spleen from recipients of lung DC– versus MLN DC–activated T cells were enumerated 24 h after one i.p. OVA challenge. (d) Thy1.2+ OTII cells in the ear skin, lung, and spleen from recipients of lung DC– versus SLN DC–activated T cells were enumerated 24 h after three daily tape strippings and epicutaneous OVA challenges. n = 12 mice per group total from three independent experiments in a and b for PPs, lung, and spleen. n = 6 mice per group total from two independent experiments in a for LP and in c and d. *, P < 0.05. Data are presented as mean (±SEM).
Figure 3.
Figure 3.
Lung-imprinting DCs are migratory DCs. Lung and TLN DCs isolated from Flt3L-expanded C57BL/6 mice 30 min or 24 h after intranasal OVA with or without intranasal CCR7 or CCR4 blocking antibody were used to activate Thy1.1+ OTII cells in vitro. DC-activated T cells were adoptively transferred into separate Thy1.2+ recipient C57BL/6 mice, followed by three inhaled OVA challenges. Thy1.1+ OTII cells in the lung from recipients of lung DC– versus TLN DC–activated T cells were enumerated when DCs were isolated either 30 min or 24 h after intranasal OVA exposure. n = 7–20 mice per group total from two to five independent experiments. *, P < 0.05; ***, P < 0.0005. Data are presented as mean (±SEM).
Figure 4.
Figure 4.
Lung DC–activated T cells home efficiently into the lung at homeostasis. DCs isolated from Flt3L-expanded C57BL/6 mice were used to activate Thy1.1+ OTII cells in vitro. DC-activated T cells were adoptively transferred into separate recipient Thy1.2+ C57BL/6 mice without OVA challenges. (a) Thy1.1+ OTII cells in the BAL and lung from recipients of DC-activated T cells were enumerated 72 h after adoptive transfer. n = 9 mice per group total from three independent experiments. (b and c) DCs were isolated from Flt3L-expanded mice. CFSE-labeled, lung DC–activated T cells and CMTMR-labeled, MLN DC– or SLN DC–activated T cells were mixed 1:1 and adoptively transferred into recipient mice without OVA challenges. (b) HI (y axis) for the lung, spleen, PPs, and SLNs (x axis) was determined 4 h after competitive adoptive transfer of lung DC– and MLN DC–activated T cells (left; n = 9 mice total from three independent experiments) and lung DC– and SLN DC–activated T cells (right; n = 9 mice total from three independent experiments). Gating on 7-AAD−CD4+ cells within the lymphocyte gate, HI was calculated as [CFSE+/CMTMR+]tissue/[CFSE+/CMTMR+]input. P-values are calculated between the HI for any tissue versus the HI for spleen. (c) CFSE+ lung DC–activated T cells (y axis) versus CMTMR+ MLN DC (x axis, top)– or SLN DC–activated T cells (x axis, bottom) were analyzed, demonstrating input cells and gated cells in the lung, spleen, PPs, and SLNs. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ****, P < 0.00005. Data are presented as mean (±SEM).
Figure 5.
Figure 5.
Expression of trafficking molecules by lung DC–activated T cells. DCs isolated from Flt3L-expanded C57BL/6 mice were used to activate OTII cells in vitro in a–e. (a) Thy1.1+ DC-activated OTII cells were either treated or untreated with PTX and adoptively transferred into Thy1.2+ recipient mice, followed by three inhaled OVA challenges. Thy1.1+ OTII cells were enumerated in the BAL and lung from recipients of PTX-treated or untreated, lung DC–activated T cells. n = 6 mice per group total from two independent experiments. (b–e) DC-activated T cells were analyzed for expression of trafficking molecules on day 5. (b) Percentage (left) and mean fluorescence intensity (MFI; right) of CCR4+ lung DC–, MLN DC–, and SLN DC–activated T cells. n = 8 independent experiments. (c) Flow cytometry of lung DC–, MLN DC–, and SLN DC–activated T cells demonstrating the expression of CD4 (x axis) versus CCR4 (top row), α4β7 (second row), CCR9 (third row), and E-selectin ligand (bottom row; y axis). (d) Percentage of α4β7+ (top), CCR9+ (middle), and E-selectin ligand+ (bottom) lung DC–, MLN DC–, and SLN DC–activated T cells. n = 6–10 separate cultures. (e) Percent expression of chemokine receptors (left) and integrins and selectin ligands (right) by lung DC–, MLN DC–, and SLN DC–activated T cells. n = 4–8 independent experiments. (f) Percent expression of chemokine receptors (left) and integrins and selectin ligands (right) by CD4+ T cells in the lymphocyte gate in lungs isolated from naive C57BL/6 mice. n = 3–10 individual lungs. *, P < 0.05; **, P < 0.005; ***, P < 0.0005; ****, P < 0.00005. Data are presented as mean (±SEM).
Figure 6.
Figure 6.
CCR4 contributes to the lung-homing advantage of lung DC–activated T cells. (a) RNA expression of CCR4 (CCL17 and CCL22), CCR6 (CCL20), and CXCR3 ligands (CXCL9 and CXCL10) in lungs of naive C57BL/6 mice. n = 8 mice. (b) Immunohistochemistry staining with CCL17 antibody (middle and right) or isotype control antibody (left) in lungs of naive mice (left and middle) and mice that received lung DC–activated OTII cells, followed by three daily inhaled OVA challenges (right), demonstrating CCL17 expression by both epithelial (top) and endothelial cells (bottom). Staining with the isotype control antibody in the lungs of mice that received OTII cells and OVA was similar to that of naive mice (not depicted). n = 13 samples from three independent experiments. Bars, 20 µm. (c) Lung HI. DCs isolated from Flt3L-expanded mice were used to activate OTII and CCR4−/− OTII cells. CFSE-labeled, lung DC–activated CCR4−/− OTII cells and CMTMR-labeled, MLN DC– or SLN DC–activated CCR4−/− OTII cells were mixed 1:1 and adoptively transferred into recipient C57BL/6 mice without OVA challenges. CFSE-labeled, lung DC–activated OTII cells and CMTMR-labeled, MLN DC– or SLN DC–activated OTII cells were also mixed 1:1 and adoptively transferred into another set of recipient C57BL/6 mice without OVA challenges. Gating on 7-AAD−CD4+ cells within the lymphocyte gate, HI for the lung at 4 h after cotransfer was calculated as [CFSE+/CMTMR+]lung/[CFSE+/CMTMR+]input. P-values are calculated between the HIs for the lung after the competitive transfer of CCR4−/− OTII cells versus OTII cells. n = 6–13 mice total from two to four independent experiments. (d) Lung DCs isolated from Flt3L-expanded mice were used to activate Thy1.2+ OTII and CCR4−/− OTII cells. Lung DC–activated OTII and CCR4−/− OTII cells were adoptively transferred into separate Thy1.1+ recipient mice, followed by three inhaled OVA challenges. Thy1.2+ OTII cells in the BAL and lung of recipient mice were enumerated 24 h after the last OVA challenge. n = 9 mice per group total from three independent experiments. *, P < 0.05; **, P < 0.005. Data are presented as mean (±SEM).
Figure 7.
Figure 7.
Lung DC–activated T cells protect against influenza. DCs isolated from Flt3L-expanded mice were used to activate Thy1.2+ OTII or CCR4−/− OTII cells in vitro. DC-activated T cells were adoptively transferred into separate Thy1.1+ C57BL/6 recipient mice. 72 h after adoptive transfer, mice were infected intranasally with 105 PFU of a live OVA323–339 expressing PR8-H1N1 influenza virus (H1ova). Lungs were harvested 72 h after infection for analysis. (a, left) Number of Thy1.2+ OTII cells in the lung from recipients of DC-activated T cells infected with H1ova. n = 5–8 mice per group in two independent experiments. P-values are calculated between recipients of lung DC-activated OTII cells versus other groups. (right) Lung flow cytometry from recipients of DC-activated T cells infected with H1ova, demonstrating Thy1.1+ (y axis) versus Thy1.2+ (x axis) cells in the CD4+ gate within the lymphocyte gate. (b) RNA expression of CCL17 and CCL22 in the lungs of mice that were infected with H1ova. n = 6–14 mice per time point in three independent experiments. (c–e) Recipients of either no cells or lung, MLN, or SLN DC–activated OTII cells or lung DC–activated CCR4−/− OTII cells were infected with H1ova as in a. n = 16–42 mice per group total from 4–11 independent experiments for c and d. n = 5–13 mice per group total from four independent experiments for e. (c) Percentage of original weight versus days after infection. (d) Percentage of surviving mice versus days after infection. (e) Viral RNA copies for the polymerase gene of the PR8 influenza virus in the lungs at day 10 after infection. *, P < 0.05; **, P < 0.005; ***, P < 0.0005. Data for a–e are presented as mean (±SEM).

References

    1. Afshar R., Strassner J.P., Seung E., Causton B., Cho J.L., Harris R.S., Hamilos D.L., Medoff B.D., Luster A.D. 2013. Compartmentalized chemokine-dependent regulatory T-cell inhibition of allergic pulmonary inflammation. J. Allergy Clin. Immunol. 131:1644–1652 10.1016/j.jaci.2013.03.002
    1. Campbell D.J., Butcher E.C. 2002. Rapid acquisition of tissue-specific homing phenotypes by CD4+ T cells activated in cutaneous or mucosal lymphoid tissues. J. Exp. Med. 195:135–141 10.1084/jem.20011502
    1. Campbell J.J., Haraldsen G., Pan J., Rottman J., Qin S., Ponath P., Andrew D.P., Warnke R., Ruffing N., Kassam N., et al. 1999. The chemokine receptor CCR4 in vascular recognition by cutaneous but not intestinal memory T cells. Nature. 400:776–780 10.1038/23495
    1. Campbell J.J., O’Connell D.J., Wurbel M.A. 2007. Cutting Edge: Chemokine receptor CCR4 is necessary for antigen-driven cutaneous accumulation of CD4 T cells under physiological conditions. J. Immunol. 178:3358–3362
    1. Centers for Disease Control and Prevention (CDC) 2013. Interim adjusted estimates of seasonal influenza vaccine effectiveness - United States, February 2013. MMWR Morb. Mortal. Wkly. Rep. 62:119–123
    1. Cho J.L., Roche M.I., Sandall B., Brass A.L., Seed B., Xavier R.J., Medoff B.D. 2012. Enhanced Tim3 activity improves survival after influenza infection. J. Immunol. 189:2879–2889 10.4049/jimmunol.1102483
    1. Chong B.F., Murphy J.E., Kupper T.S., Fuhlbrigge R.C. 2004. E-selectin, thymus- and activation-regulated chemokine/CCL17, and intercellular adhesion molecule-1 are constitutively coexpressed in dermal microvessels: a foundation for a cutaneous immunosurveillance system. J. Immunol. 172:1575–1581
    1. Chvatchko Y., Hoogewerf A.J., Meyer A., Alouani S., Juillard P., Buser R., Conquet F., Proudfoot A.E., Wells T.N., Power C.A. 2000. A key role for CC chemokine receptor 4 in lipopolysaccharide-induced endotoxic shock. J. Exp. Med. 191:1755–1764 10.1084/jem.191.10.1755
    1. Ciabattini A., Pettini E., Fiorino F., Prota G., Pozzi G., Medaglini D. 2011. Distribution of primed T cells and antigen-loaded antigen presenting cells following intranasal immunization in mice. PLoS ONE. 6:e19346 10.1371/journal.pone.0019346
    1. Clark R.A., Chong B., Mirchandani N., Brinster N.K., Yamanaka K., Dowgiert R.K., Kupper T.S. 2006. The vast majority of CLA+ T cells are resident in normal skin. J. Immunol. 176:4431–4439
    1. Dudda J.C., Simon J.C., Martin S. 2004. Dendritic cell immunization route determines CD8+ T cell trafficking to inflamed skin: role for tissue microenvironment and dendritic cells in establishment of T cell-homing subsets. J. Immunol. 172:857–863
    1. Fainaru O., Shseyov D., Hantisteanu S., Groner Y. 2005. Accelerated chemokine receptor 7-mediated dendritic cell migration in Runx3 knockout mice and the spontaneous development of asthma-like disease. Proc. Natl. Acad. Sci. USA. 102:10598–10603 10.1073/pnas.0504787102
    1. Freeman C.M., Stolberg V.R., Chiu B.C., Lukacs N.W., Kunkel S.L., Chensue S.W. 2006. CCR4 participation in Th type 1 (mycobacterial) and Th type 2 (schistosomal) anamnestic pulmonary granulomatous responses. J. Immunol. 177:4149–4158
    1. Gretz J.E., Norbury C.C., Anderson A.O., Proudfoot A.E., Shaw S. 2000. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J. Exp. Med. 192:1425–1440 10.1084/jem.192.10.1425
    1. Hamada H., Bassity E., Flies A., Strutt T.M., Garcia-Hernandez Mde.L., McKinstry K.K., Zou T., Swain S.L., Dutton R.W. 2013. Multiple redundant effector mechanisms of CD8+ T cells protect against influenza infection. J. Immunol. 190:296–306 10.4049/jimmunol.1200571
    1. Hao X., Kim T.S., Braciale T.J. 2008. Differential response of respiratory dendritic cell subsets to influenza virus infection. J. Virol. 82:4908–4919 10.1128/JVI.02367-07
    1. Hartl D., Griese M., Kappler M., Zissel G., Reinhardt D., Rebhan C., Schendel D.J., Krauss-Etschmann S. 2006. Pulmonary T(H)2 response in Pseudomonas aeruginosa-infected patients with cystic fibrosis. J. Allergy Clin. Immunol. 117:204–211 10.1016/j.jaci.2005.09.023
    1. Helft J., Merad M. 2010. Isolation of cutaneous dendritic cells. Methods Mol. Biol. 595:231–233 10.1007/978-1-60761-421-0_15
    1. Hoft D.F., Babusis E., Worku S., Spencer C.T., Lottenbach K., Truscott S.M., Abate G., Sakala I.G., Edwards K.M., Creech C.B., et al. 2011. Live and inactivated influenza vaccines induce similar humoral responses, but only live vaccines induce diverse T-cell responses in young children. J. Infect. Dis. 204:845–853 10.1093/infdis/jir436
    1. Iwata M., Hirakiyama A., Eshima Y., Kagechika H., Kato C., Song S.Y. 2004. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 21:527–538 10.1016/j.immuni.2004.08.011
    1. Jakubzick C., Bogunovic M., Bonito A.J., Kuan E.L., Merad M., Randolph G.J. 2008. Lymph-migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph nodes. J. Exp. Med. 205:2839–2850 10.1084/jem.20081430
    1. Jiang X., Clark R.A., Liu L., Wagers A.J., Fuhlbrigge R.C., Kupper T.S. 2012. Skin infection generates non-migratory memory CD8+ T(RM) cells providing global skin immunity. Nature. 483:227–231 10.1038/nature10851
    1. Johansson-Lindbom B., Svensson M., Wurbel M.A., Malissen B., Márquez G., Agace W. 2003. Selective generation of gut tropic T cells in gut-associated lymphoid tissue (GALT): requirement for GALT dendritic cells and adjuvant. J. Exp. Med. 198:963–969 10.1084/jem.20031244
    1. Kallinich T., Schmidt S., Hamelmann E., Fischer A., Qin S., Luttmann W., Virchow J.C., Kroczek R.A. 2005. Chemokine-receptor expression on T cells in lung compartments of challenged asthmatic patients. Clin. Exp. Allergy. 35:26–33 10.1111/j.1365-2222.2004.02132.x
    1. Kawasaki S., Takizawa H., Yoneyama H., Nakayama T., Fujisawa R., Izumizaki M., Imai T., Yoshie O., Homma I., Yamamoto K., Matsushima K. 2001. Intervention of thymus and activation-regulated chemokine attenuates the development of allergic airway inflammation and hyperresponsiveness in mice. J. Immunol. 166:2055–2062
    1. Koelle D.M., Liu Z., McClurkan C.M., Topp M.S., Riddell S.R., Pamer E.G., Johnson A.S., Wald A., Corey L. 2002. Expression of cutaneous lymphocyte-associated antigen by CD8(+) T cells specific for a skin-tropic virus. J. Clin. Invest. 110:537–548
    1. Kunkel E.J., Boisvert J., Murphy K., Vierra M.A., Genovese M.C., Wardlaw A.J., Greenberg H.B., Hodge M.R., Wu L., Butcher E.C., Campbell J.J. 2002. Expression of the chemokine receptors CCR4, CCR5, and CXCR3 by human tissue-infiltrating lymphocytes. Am. J. Pathol. 160:347–355 10.1016/S0002-9440(10)64378-7
    1. Lambrecht B.N., Hammad H. 2012. Lung dendritic cells in respiratory viral infection and asthma: from protection to immunopathology. Annu. Rev. Immunol. 30:243–270 10.1146/annurev-immunol-020711-075021
    1. Lee H.K., Zamora M., Linehan M.M., Iijima N., Gonzalez D., Haberman A., Iwasaki A. 2009. Differential roles of migratory and resident DCs in T cell priming after mucosal or skin HSV-1 infection. J. Exp. Med. 206:359–370 10.1084/jem.20080601
    1. Liu L., Fuhlbrigge R.C., Karibian K., Tian T., Kupper T.S. 2006. Dynamic programming of CD8+ T cell trafficking after live viral immunization. Immunity. 25:511–520 10.1016/j.immuni.2006.06.019
    1. Liu L., Zhong Q., Tian T., Dubin K., Athale S.K., Kupper T.S. 2010. Epidermal injury and infection during poxvirus immunization is crucial for the generation of highly protective T cell-mediated immunity. Nat. Med. 16:224–227 10.1038/nm.2078
    1. Lloyd C.M., Delaney T., Nguyen T., Tian J., Martinez-A C., Coyle A.J., Gutierrez-Ramos J.C. 2000. CC chemokine receptor (CCR)3/eotaxin is followed by CCR4/monocyte-derived chemokine in mediating pulmonary T helper lymphocyte type 2 recruitment after serial antigen challenge in vivo. J. Exp. Med. 191:265–274 10.1084/jem.191.2.265
    1. Mach N., Gillessen S., Wilson S.B., Sheehan C., Mihm M., Dranoff G. 2000. Differences in dendritic cells stimulated in vivo by tumors engineered to secrete granulocyte-macrophage colony-stimulating factor or Flt3-ligand. Cancer Res. 60:3239–3246
    1. Masopust D., Vezys V., Marzo A.L., Lefrançois L. 2001. Preferential localization of effector memory cells in nonlymphoid tissue. Science. 291:2413–2417 10.1126/science.1058867
    1. Masopust D., Vezys V., Usherwood E.J., Cauley L.S., Olson S., Marzo A.L., Ward R.L., Woodland D.L., Lefrançois L. 2004. Activated primary and memory CD8 T cells migrate to nonlymphoid tissues regardless of site of activation or tissue of origin. J. Immunol. 172:4875–4882
    1. Masopust D., Choo D., Vezys V., Wherry E.J., Duraiswamy J., Akondy R., Wang J., Casey K.A., Barber D.L., Kawamura K.S., et al. 2010. Dynamic T cell migration program provides resident memory within intestinal epithelium. J. Exp. Med. 207:553–564 10.1084/jem.20090858
    1. McCully M.L., Ladell K., Hakobyan S., Mansel R.E., Price D.A., Moser B. 2012. Epidermis instructs skin homing receptor expression in human T cells. Blood. 120:4591–4598 10.1182/blood-2012-05-433037
    1. Mikhak Z., Fleming C.M., Medoff B.D., Thomas S.Y., Tager A.M., Campanella G.S., Luster A.D. 2006. STAT1 in peripheral tissue differentially regulates homing of antigen-specific Th1 and Th2 cells. J. Immunol. 176:4959–4967
    1. Mikhak Z., Fukui M., Farsidjani A., Medoff B.D., Tager A.M., Luster A.D. 2009. Contribution of CCR4 and CCR8 to antigen-specific T(H)2 cell trafficking in allergic pulmonary inflammation. J. Allergy Clin. Immunol. 123:67–73 10.1016/j.jaci.2008.09.049
    1. Mora J.R., Bono M.R., Manjunath N., Weninger W., Cavanagh L.L., Rosemblatt M., Von Andrian U.H. 2003. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature. 424:88–93 10.1038/nature01726
    1. Mora J.R., Cheng G., Picarella D., Briskin M., Buchanan N., von Andrian U.H. 2005. Reciprocal and dynamic control of CD8 T cell homing by dendritic cells from skin- and gut-associated lymphoid tissues. J. Exp. Med. 201:303–316 10.1084/jem.20041645
    1. Olkhanud P.B., Baatar D., Bodogai M., Hakim F., Gress R., Anderson R.L., Deng J., Xu M., Briest S., Biragyn A. 2009. Breast cancer lung metastasis requires expression of chemokine receptor CCR4 and regulatory T cells. Cancer Res. 69:5996–6004 10.1158/0008-5472.CAN-08-4619
    1. Osterholm M.T., Kelley N.S., Sommer A., Belongia E.A. 2012. Efficacy and effectiveness of influenza vaccines: a systematic review and meta-analysis. Lancet Infect. Dis. 12:36–44 10.1016/S1473-3099(11)70295-X
    1. Oyoshi M.K., Elkhal A., Scott J.E., Wurbel M.A., Hornick J.L., Campbell J.J., Geha R.S. 2011. Epicutaneous challenge of orally immunized mice redirects antigen-specific gut-homing T cells to the skin. J. Clin. Invest. 121:2210–2220 10.1172/JCI43586
    1. Purwar R., Campbell J., Murphy G., Richards W.G., Clark R.A., Kupper T.S. 2011. Resident memory T cells (T(RM)) are abundant in human lung: diversity, function, and antigen specificity. PLoS ONE. 6:e16245 10.1371/journal.pone.0016245
    1. Reinhardt R.L., Kang S.J., Liang H.E., Locksley R.M. 2006. T helper cell effector fates—who, how and where? Curr. Opin. Immunol. 18:271–277 10.1016/j.coi.2006.03.003
    1. Reiss Y., Proudfoot A.E., Power C.A., Campbell J.J., Butcher E.C. 2001. CC chemokine receptor (CCR)4 and the CCR10 ligand cutaneous T cell–attracting chemokine (CTACK) in lymphocyte trafficking to inflamed skin. J. Exp. Med. 194:1541–1547 10.1084/jem.194.10.1541
    1. Rott L.S., Rosé J.R., Bass D., Williams M.B., Greenberg H.B., Butcher E.C. 1997. Expression of mucosal homing receptor alpha4beta7 by circulating CD4+ cells with memory for intestinal rotavirus. J. Clin. Invest. 100:1204–1208 10.1172/JCI119633
    1. Sather B.D., Treuting P., Perdue N., Miazgowicz M., Fontenot J.D., Rudensky A.Y., Campbell D.J. 2007. Altering the distribution of Foxp3+ regulatory T cells results in tissue-specific inflammatory disease. J. Exp. Med. 204:1335–1347 10.1084/jem.20070081
    1. Shang L., Thirunarayanan N., Viejo-Borbolla A., Martin A.P., Bogunovic M., Marchesi F., Unkeless J.C., Ho Y., Furtado G.C., Alcami A., et al. 2009. Expression of the chemokine binding protein M3 promotes marked changes in the accumulation of specific leukocytes subsets within the intestine. Gastroenterology. 137:1006–1018 10.1053/j.gastro.2009.05.055
    1. Shankar S.P., Wilson M.S., DiVietro J.A., Mentink-Kane M.M., Xie Z., Wynn T.A., Druey K.M. 2012. RGS16 attenuates pulmonary Th2/Th17 inflammatory responses. J. Immunol. 188:6347–6356 10.4049/jimmunol.1103781
    1. Sigmundsdottir H., Pan J., Debes G.F., Alt C., Habtezion A., Soler D., Butcher E.C. 2007. DCs metabolize sunlight-induced vitamin D3 to ‘program’ T cell attraction to the epidermal chemokine CCL27. Nat. Immunol. 8:285–293 10.1038/ni1433
    1. Soler D., Humphreys T.L., Spinola S.M., Campbell J.J. 2003. CCR4 versus CCR10 in human cutaneous TH lymphocyte trafficking. Blood. 101:1677–1682 10.1182/blood-2002-07-2348
    1. Stagg A.J., Kamm M.A., Knight S.C. 2002. Intestinal dendritic cells increase T cell expression of alpha4beta7 integrin. Eur. J. Immunol. 32:1445–1454 10.1002/1521-4141(200205)32:5<1445::AID-IMMU1445>;2-E
    1. Stolberg V.R., Chiu B.C., Schmidt B.M., Kunkel S.L., Sandor M., Chensue S.W. 2011. CC chemokine receptor 4 contributes to innate NK and chronic stage T helper cell recall responses during Mycobacterium bovis infection. Am. J. Pathol. 178:233–244 10.1016/j.ajpath.2010.11.036
    1. Strutt T.M., McKinstry K.K., Dibble J.P., Winchell C., Kuang Y., Curtis J.D., Huston G., Dutton R.W., Swain S.L. 2010. Memory CD4+ T cells induce innate responses independently of pathogen. Nat. Med. 16:558–564 10.1038/nm.2142
    1. Teijaro J.R., Turner D., Pham Q., Wherry E.J., Lefrançois L., Farber D.L. 2011. Cutting edge: Tissue-retentive lung memory CD4 T cells mediate optimal protection to respiratory virus infection. J. Immunol. 187:5510–5514 10.4049/jimmunol.1102243
    1. Thomas P.G., Brown S.A., Yue W., So J., Webby R.J., Doherty P.C. 2006. An unexpected antibody response to an engineered influenza virus modifies CD8+ T cell responses. Proc. Natl. Acad. Sci. USA. 103:2764–2769 10.1073/pnas.0511185103
    1. Thomas S.Y., Banerji A., Medoff B.D., Lilly C.M., Luster A.D. 2007. Multiple chemokine receptors, including CCR6 and CXCR3, regulate antigen-induced T cell homing to the human asthmatic airway. J. Immunol. 179:1901–1912
    1. Vermaelen K.Y., Carro-Muino I., Lambrecht B.N., Pauwels R.A. 2001. Specific migratory dendritic cells rapidly transport antigen from the airways to the thoracic lymph nodes. J. Exp. Med. 193:51–60 10.1084/jem.193.1.51
    1. von Wulffen W., Steinmueller M., Herold S., Marsh L.M., Bulau P., Seeger W., Welte T., Lohmeyer J., Maus U.A. 2007. Lung dendritic cells elicited by Fms-like tyrosine 3-kinase ligand amplify the lung inflammatory response to lipopolysaccharide. Am. J. Respir. Crit. Care Med. 176:892–901 10.1164/rccm.200608-1068OC

Source: PubMed

3
订阅